Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(31): e2308750120, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37487068

RESUMEN

Adipose tissue is central to regulation of energy homeostasis. Adaptive thermogenesis, which relies on mitochondrial oxidative phosphorylation (Ox-Phos), dissipates energy to counteract obesity. On the other hand, chronic inflammation in adipose tissue is linked to type 2 diabetes and obesity. Here, we show that nuclear factor I-A (NFIA), a transcriptional regulator of brown and beige adipocytes, improves glucose homeostasis by upregulation of Ox-Phos and reciprocal downregulation of inflammation. Mice with transgenic expression of NFIA in adipocytes exhibited improved glucose tolerance and limited weight gain. NFIA up-regulates Ox-Phos and brown-fat-specific genes by enhancer activation that involves facilitated genomic binding of PPARγ. In contrast, NFIA in adipocytes, but not in macrophages, down-regulates proinflammatory cytokine genes to ameliorate adipose tissue inflammation. NFIA binds to regulatory region of the Ccl2 gene, which encodes proinflammatory cytokine MCP-1 (monocyte chemoattractant protein-1), to down-regulate its transcription. CCL2 expression was negatively correlated with NFIA expression in human adipose tissue. These results reveal the beneficial effect of NFIA on glucose and body weight homeostasis and also highlight previously unappreciated role of NFIA in suppressing adipose tissue inflammation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Factores de Transcripción NFI , Humanos , Animales , Ratones , Adipocitos , Homeostasis , Inflamación , Tejido Adiposo Pardo , Citocinas
2.
J Genet Couns ; 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37965685

RESUMEN

Patients with Marfan syndrome (MFS) present with various symptoms, such as aortic aneurysm/dissection, tall stature, and lens deviation. Among them, acute aortic dissection is a complication that leads to sudden death. Some individuals with MFS are reluctant to see a cardiologist and discontinue regular checkups until they develop life-threatening complications. We conducted a grounded theory study to investigate how individuals with MFS decided whether to adhere to healthcare recommendations, specifically to attend cardiology appointments. The study recruited individuals with a clinical or genetic diagnosis of MFS from a Japanese university hospital and individuals from a support group. Semi-structured interviews were conducted with 28 consenting participants. In this study, we identified the decision-making processes of individuals with MFS concerning their cardiology visits. We extracted "perception of the gap between their health status and medical recommendations" as the central category. This decision-making process consisted of three parts: (A) the process by which an individual with MFS sees a cardiologist for the first time, (B) the process by which an individual with MFS keeps up with cardiology checkups, and (C) the process by which parents bring their children with MFS to the cardiologist. Individuals who learned of the possibility of MFS decided whether to adhere to medical recommendations depending on how they perceived the gap between their health status and the medical recommendations. In addition to medical information and treatment experience, adaptation to MFS, which changed through interactions with others, influenced the perception of the gap. This study suggests the role of genetic counseling and molecular genetic diagnosis as factors that may facilitate adaptation to MFS. The involvement of genetic counselors is important for helping individuals with MFS keep up with regular checkups while affirming their own experiences. These results provide insight into adherence to medical recommendations for individuals with MFS.

3.
PLoS Genet ; 16(9): e1009044, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32991581

RESUMEN

The transcription factor nuclear factor I-A (NFIA) is a regulator of brown adipocyte differentiation. Here we show that the C-terminal 17 amino acid residues of NFIA (which we call pro#3 domain) are required for the transcriptional activity of NFIA. Full-length NFIA-but not deletion mutant lacking pro#3 domain-rescued impaired expression of PPARγ, the master transcriptional regulator of adipogenesis and impaired adipocyte differentiation in NFIA-knockout cells. Mechanistically, the ability of NFIA to penetrate chromatin and bind to the crucial Pparg enhancer is mediated through pro#3 domain. However, the deletion mutant still binds to Myod1 enhancer to repress expression of MyoD, the master transcriptional regulator of myogenesis as well as proximally transcribed non-coding RNA called DRReRNA, via competition with KLF5 in terms of enhancer binding, leading to suppression of myogenic gene program. Therefore, the negative effect of NFIA on the myogenic gene program is, at least partly, independent of the positive effect on PPARγ expression and its downstream adipogenic gene program. These results uncover multiple ways of action of NFIA to ensure optimal regulation of brown and beige adipocyte differentiation.


Asunto(s)
Adipocitos Beige/citología , Adipocitos Marrones/citología , Adipogénesis/fisiología , Desarrollo de Músculos/fisiología , Factores de Transcripción NFI/metabolismo , Adipocitos Beige/fisiología , Adipocitos Marrones/fisiología , Adipogénesis/genética , Animales , Diferenciación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos/genética , Proteína MioD/genética , Miogenina/genética , Factores de Transcripción NFI/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Prolina , Dominios Proteicos
4.
Cytogenet Genome Res ; 162(11-12): 625-631, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37245502

RESUMEN

Transient abnormal myelopoiesis (TAM) is a unique neonatal leukemoid reaction caused by a pathognomonic GATA1 mutation in conjunction with the gene dosage effect of trisomy 21, which is either of germline or somatic origin. We encountered a 48,XYY,+21 phenotypically normal neonate with Down syndrome who developed TAM due to cryptic germline mosaicism. Quantification of the mosaic ratio was complicated by an overestimation bias of hyperproliferating TAM within the germline component. To establish a workflow for such a clinical scenario, we analyzed the cytogenetic findings of neonates with TAM associated with somatic or low-level germline mosaicism. We showed that multistep diagnostic procedures (i.e., paired cytogenetic analyses of peripheral blood specimens in culture with or without phytohemagglutinin; serial cytogenetic studies of more than one tissue, such as the buccal membrane; and complementary DNA-based GATA1 mutation screening) can verify the specificity of cytogenetic testing for phenotypically normal neonates with TAM suspected of mosaicism.

5.
J Hum Genet ; 67(11): 623-628, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35787654

RESUMEN

Although very low birth weight (VLBW) is well studied in neonatology and the perinatal prognosis of VLBW infants has improved over time, little is known about the prognosis of VLBW infants with trisomy 21 (T21). We aimed to investigate the mortality and morbidity of VLBW infants with T21 during NICU admission in Japan, in comparison to those of infants without birth defects (BD-). Maternal and neonatal data of infants weighing 1500 grams or less admitted to the centers of the Neonatal Research Network of Japan from 2003 to 2016 were collected prospectively. Of 60,136 infants, 328 (0.55%) had T21. Although maternal age in the case of T21 infants was higher, maternal complications tended to be less frequent than in those with BD-. Multivariable analysis revealed that morbidities were higher in infants with T21 than in those with BD- but respiratory distress syndrome and retinopathy of prematurity were less frequent in those with T21 (p < 0.001, and p = 0.014, respectively), and no significant difference was observed between the two groups in the proportion of late-onset circulatory collapse of prematurity as well as cystic periventricular leukomalacia (p = 0.739 and p = 0.733, respectively). The survival rate at discharge from the NICU was 77% and 94% for T21 and BD-, respectively. This was the first nationwide survey of VLBW infants with T21 in Japan. Although there were no data regarding the timing of diagnosis, these data will aid prenatal genetic counseling and perinatal management of T21 infants.


Asunto(s)
Síndrome de Down , Recién Nacido , Lactante , Embarazo , Femenino , Humanos , Síndrome de Down/epidemiología , Japón/epidemiología , Recién Nacido de muy Bajo Peso , Morbilidad , Trisomía
6.
Circ J ; 87(1): 123-129, 2022 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-36372399

RESUMEN

BACKGROUND: We assessed the awareness of multidisciplinary healthcare professionals of the challenges related to implementation of molecular autopsy (MA) for sudden cardiac death (SCD) among children and young adults.Methods and Results: We conducted 11 focus groups with 31 multidisciplinary healthcare professionals, and categorized them into 2 themes: values, and challenges of MA implementation. The participants recognized 2 different values of MA: discovering the unknown cause of SCD, and SCD prevention among family members of victims. The coexistence of these values makes the MA process and role of professionals more complex. Participants were concerned about the psychological burden for bereaved family members and mentioned challenges in each process of the MA delivery system: obtaining consent, cause of death investigation, disclosing results, and preventive intervention. CONCLUSIONS: MA is a valuable procedure both in terms of forensic and preventive medicine. However, the dual meanings and complex characteristics of genetic information is a potential source of concern and confusion among healthcare professionals as well as bereaved family members. Increasing awareness among healthcare professionals of the MA process is essential for connecting all related areas of expertise.


Asunto(s)
Muerte Súbita Cardíaca , Niño , Adulto Joven , Humanos , Grupos Focales , Autopsia/métodos , Japón , Muerte Súbita Cardíaca/prevención & control , Muerte Súbita Cardíaca/etiología , Causas de Muerte
7.
J Hum Genet ; 66(3): 273-285, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32943740

RESUMEN

Trisomy 18 (T18) and trisomy 13 (T13) are major concerns in prenatal genetic testing due to their poor prognosis; very low birth weight (VLBW) is also a concern in neonatology. The aim of this study was to investigate the mortality and morbidity of VLBW infants diagnosed with T18/T13 in Japan, compared with those with no birth defects (BD-). Maternal and neonatal data were collected prospectively from infants weighing <1501 g and were admitted to centers of the Neonatal Research Network of Japan during 2003 to 2016. Among 60,136 infants, 563 and 60 was diagnosed with T18 and T13, respectively. Although the age of mothers of infants with T18/T13 was higher, the frequency of maternal complications was lower than those with BD-. With maternal and neonatal characteristic adjustments, T18/T13 had a higher incidence of each morbidity when compared with BD-. Mortality rates in the NICU were 70, 77, and 5.8% for T18, T13, and BD-, respectively, while the survival discharge rates of T18 and T13 were 29.5 and 23.3%, respectively, which was significantly higher than previous reports. This was the first nationwide survey for VLBW infants with T18/T13 in Japan; this novel data will be relevant and useful for prenatal genetic counseling and perinatal management. Although T18/T13 were considered to be fatal in the past, with proper epidemiological information, discussions with affected families, and compassionate patient care, the mortality rate of T18/T13 can be improved.


Asunto(s)
Enfermedades del Recién Nacido/epidemiología , Recién Nacido de muy Bajo Peso , Síndrome de la Trisomía 13/epidemiología , Síndrome de la Trisomía 18/epidemiología , Anomalías Múltiples/epidemiología , Anomalías Múltiples/genética , Pueblo Asiatico/genética , Corioamnionitis/epidemiología , Comorbilidad , Susceptibilidad a Enfermedades , Femenino , Rotura Prematura de Membranas Fetales/epidemiología , Humanos , Incidencia , Recién Nacido , Enfermedades del Prematuro/epidemiología , Unidades de Cuidado Intensivo Neonatal , Japón/epidemiología , Masculino , Edad Materna , Embarazo , Complicaciones del Embarazo/epidemiología , Pronóstico , Estudios Prospectivos , Síndrome de la Trisomía 13/etnología , Síndrome de la Trisomía 13/mortalidad , Síndrome de la Trisomía 18/etnología , Síndrome de la Trisomía 18/mortalidad
8.
Biochem Biophys Res Commun ; 531(1): 51-55, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31980177

RESUMEN

Genomic regions with guanine (G)-rich sequences make non-Watson-Crick base pairs, which result in the formation of unique nucleic acid structures called G-quadruplexes (G4s) in cells. Studies have suggested that abnormal G4s are involved in neurological diseases. For example, the formation of G4s caused by expansion of G-rich sequences is implicated in C9orf72-mediated amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD), and fragile X-related tremor/ataxia syndrome (FXTAS). In addition, the disruption and/or mutation of G4 binding proteins (G4BPs), such as heterogeneous nuclear ribonucleoproteins (hnRNPs) and DNA/RNA helicases, is related to neurological diseases. For instance, mutations in a G4BP called ATRX lead to a neurodevelopmental disorder, ATR-X syndrome, which is associated with intellectual disability. We found that porphyrins are potential candidate drugs for treating ATR-X syndrome through their G4 binding ability. Importantly, intracellular porphyrins are produced from 5-aminolevulinic acid (5-ALA) in vivo. Oral administration of 5-ALA improved cognitive dysfunction in an ATR-X syndrome model mouse, and language ability in an ATR-X syndrome patient. In this review, we suggest a novel therapeutic strategy targeting G4s using porphyrins in neurological diseases.


Asunto(s)
G-Cuádruplex/efectos de los fármacos , Discapacidad Intelectual Ligada al Cromosoma X/tratamiento farmacológico , Porfirinas/farmacología , Talasemia alfa/tratamiento farmacológico , Animales , Descubrimiento de Drogas/métodos , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/genética , Terapia Molecular Dirigida/métodos , Porfirinas/química , Talasemia alfa/genética
9.
J Pediatr ; 226: 106-111.e10, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32634406

RESUMEN

OBJECTIVE: To evaluate the mortality and morbidity of very low birth weight (VLBW) preterm infants with birth defects in Japan. STUDY DESIGN: Data were collected prospectively for infants weighing <1501 g and born at <37 weeks of gestation admitted to centers of the Neonatal Research Network of Japan during 2003-2016. We compared outcomes of infants with and without birth defects using Pearson χ2 test, Wilcoxon rank-sum test, log-rank test, nominal logistic regression analysis, and stratified analysis by birth defect subgroups. This study was approved by the Ethics Committee of Kyoto University Graduate School and Faculty of Medicine. RESULTS: Among 57 730 VLBW preterm infants, 3557 infants (6.2%) were born with birth defects. Chromosomal abnormalities, congenital heart defects, and congenital malformation of the digestive system were the most common categories. Among diseases, Trisomy 18, Down syndrome, and cleft palate were the most prevalent. There were significant differences between perinatal characteristics of infants with and without birth defects. Most categories of morbidity occurred more frequently in infants with birth defects compared with those without birth defects. The aOR for mortality during the neonatal intensive care unit admission was 10.6 (95% CI 9.5-11.7) for infants with birth defects. A stratified analysis identified birth defect categories with good, moderate, and poor prognoses. CONCLUSIONS: This detailed information about mortality and morbidity of preterm VLBW infants with birth defects should be useful for genetic counseling as well as prenatal and neonatal care, with the limitation that we lacked information about the timing of diagnosis, abortion, or stillbirth.


Asunto(s)
Anomalías Congénitas/epidemiología , Enfermedades del Prematuro/epidemiología , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Cuidado Intensivo Neonatal , Japón/epidemiología , Masculino , Tasa de Supervivencia
10.
J Hum Genet ; 65(3): 337-343, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31857675

RESUMEN

Although alkaline phosphatase (ALP) activity is relatively low in carriers of recessive type hypophosphatasia (HPP), most are asymptomatic and therefore do not undergo medical evaluations. We analyzed the association of ALP-encoding ALPL variants with serum ALP and bone traits in the general Japanese population. Study participants (n = 9671) were from the Nagahama Study, which was a longitudinal cohort study of an apparently healthy general Japanese population. ALPL variants were analyzed by whole-genome sequencing or TaqMan probe assays using DNA extracted from peripheral blood samples. The speed of sound in calcaneal bone was assessed by quantitative ultrasound (QUS) and used as surrogate measures of bone mineral density. We identified 13 ALPL variants. Minor allele frequencies of three variants were higher than expected. Variant c.529G > A has been reported as a possible pathogenic variant for adult type HPP. Variants c.979C > T and c.1559delT are reported as pathogenic variants for perinatal severe HPP or infantile HPP. The allele frequencies of c.529G > A, c.979C > T, and c.1559delT were 0.0107, 0.0040, and 0.0014, respectively. Serum ALP activity was significantly lower and differed among the three variants (P < 0.001), as well as between individuals with and without any of the three variants (P < 0.001). Serum ALP activity was inversely associated with QUS values, although no direct association was observed between the ALPL variants and QUS values. An association between serum ALP activity and QUS was confirmed; however, we failed to detect an association between ALPL variants and bone traits in the general Japanese population.


Asunto(s)
Fosfatasa Alcalina/genética , Densidad Ósea/genética , Desarrollo Óseo/genética , Predisposición Genética a la Enfermedad , Adulto , Fosfatasa Alcalina/sangre , Huesos/metabolismo , Huesos/patología , Análisis Mutacional de ADN , Femenino , Humanos , Hipofosfatasia/sangre , Hipofosfatasia/epidemiología , Hipofosfatasia/genética , Japón/epidemiología , Estudios Longitudinales , Masculino , Fenotipo , Embarazo , Secuenciación Completa del Genoma
11.
Pharm Res ; 37(3): 61, 2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32124083

RESUMEN

PURPOSE: Cyclocreatine, a creatine analog, is a candidate drug for treating patients with cerebral creatine deficiency syndromes (CCDSs) caused by creatine transporter (CRT, SLC6A8) deficiency, which reduces brain creatine level. The purpose of this study was to clarify the characteristics of cyclocreatine transport in HEK293 cells, which highly express endogenous CRT, in hCMEC/D3 cells, a human blood-brain barrier (BBB) model, and in CCDSs patient-derived fibroblasts with CRT mutations. METHODS: Cells were incubated at 37°C with [14C]cyclocreatine (9 µM) and [14C]creatine (9 µM) for specified periods of times in the presence or absence of inhibitors, while the siRNAs were transfected by lipofection. Protein expression and mRNA expression were quantified using targeted proteomics and quantitative PCR, respectively. RESULTS: [14C]Cyclocreatine was taken up by HEK293 cells in a time-dependent manner, while exhibiting saturable kinetics. The inhibition and siRNA knockdown studies demonstrated that the uptake of [14C]cyclocreatine by both HEK293 and hCMEC/D3 cells was mediated predominantly by CRT as well as [14C]creatine. In addition, uptake of [14C]cyclocreatine and [14C]creatine by the CCDSs patient-derived fibroblasts was found to be largely reduced. CONCLUSION: The present study suggests that cyclocreatine is a CRT substrate, where CRT is the predominant contributor to influx of cyclocreatine into the brain at the BBB. Our findings provide vital insights for the purposes of treating CCDSs patients using cyclocreatine.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Creatina/deficiencia , Creatinina/análogos & derivados , Fibroblastos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transporte Biológico , Barrera Hematoencefálica/citología , Línea Celular , Células Cultivadas , Creatina/metabolismo , Creatinina/metabolismo , Creatinina/farmacocinética , Células HEK293 , Humanos
13.
Biol Pharm Bull ; 40(1): 49-55, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28049948

RESUMEN

Cerebral creatine deficiency syndromes (CCDSs) are caused by loss-of-function mutations in creatine transporter (CRT, SLC6A8), which transports creatine at the blood-brain barrier and into neurons of the central nervous system (CNS). This results in low cerebral creatine levels, and patients exhibit mental retardation, poor language skills and epilepsy. We identified a novel human CRT gene missense mutation (c.1681 G>C, G561R) in Japanese CCDSs patients. The purpose of the present study was to evaluate the reduction of creatine transport in G561R-mutant CRT-expressing 293 cells, and to clarify the mechanism of its functional attenuation. G561R-mutant CRT exhibited greatly reduced creatine transport activity compared to wild-type CRT (WT-CRT) when expressed in 293 cells. Also, the mutant protein is localized mainly in intracellular membrane fraction, while WT-CRT is localized in plasma membrane. Western blot analysis revealed a 68 kDa band of WT-CRT protein in plasma membrane fraction, while G561R-mutant CRT protein predominantly showed bands at 55, 110 and 165 kDa in crude membrane fraction. The bands of both WT-CRT and G561R-mutant CRT were shifted to 50 kDa by N-glycosidase treatment. Our results suggest that the functional impairment of G561R-mutant CRT was probably caused by incomplete N-linked glycosylation due to misfolding during protein maturation, leading to oligomer formation and changes of cellular localization.


Asunto(s)
Creatina/metabolismo , Proteínas de Transporte de Membrana/genética , Transporte Biológico , Encéfalo/metabolismo , Membrana Celular/metabolismo , Creatina/deficiencia , Glicosilación , Células HEK293 , Humanos , Proteínas de Transporte de Membrana/metabolismo , Mutación Missense , Síndrome
14.
Chembiochem ; 17(10): 928-35, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-26991472

RESUMEN

ATR-X (α-thalassemia/mental retardation X-linked) syndrome is caused by mutations in chromatin remodeler ATRX. ATRX can bind the variable number of tandem repeats (VNTR) sequence in the promoter region of the α-globin gene cluster. The VNTR sequence, which contains the potential G-quadruplex-forming sequence CGC(GGGGCGGGG)n , is involved in the downregulation of α-globin expression. We investigated G-quadruplex and i-motif formation in single-stranded DNA and long double-stranded DNA. The promoter region without the VNTR sequence showed approximately twofold higher luciferase activity than the promoter region harboring the VNTR sequence. G-quadruplex stabilizers hemin and TMPyP4 reduced the luciferase activity, whereas expression of ATRX led to a recovery in reporter activity. Our results demonstrate that stable G-quadruplex formation by the VNTR sequence downregulates the expression of α-globin genes and that ATRX might bind to and resolve the G-quadruplex.


Asunto(s)
ADN Helicasas/metabolismo , G-Cuádruplex , Regulación de la Expresión Génica/genética , Repeticiones de Minisatélite/genética , Globinas alfa/genética , Secuencia de Bases , Dicroismo Circular , Regulación hacia Abajo , Genes Reporteros , Hemina/química , Hemina/metabolismo , Humanos , Microscopía de Fuerza Atómica , Porfirinas/química , Porfirinas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Globinas alfa/metabolismo
15.
Am J Med Genet A ; 167(6): 1349-53, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25900396

RESUMEN

Next-generation sequencing has enabled the screening for a causative mutation in X-linked intellectual disability (XLID). We identified KIAA2022 mutations in two unrelated male patients by targeted sequencing. We selected 13 Japanese male patients with severe intellectual disability (ID), including four sibling patients and nine sporadic patients. Two of thirteen had a KIAA2022 mutation. Patient 1 was a 3-year-old boy. He had severe ID with autistic behavior and hypotonia. Patient 2 was a 5-year-old boy. He also had severe ID with autistic behavior, hypotonia, central hypothyroidism, and steroid-dependent nephrotic syndrome. Both patients revealed consistent distinctive features, including upswept hair, narrow forehead, downslanting eyebrows, wide palpebral fissures, long nose, hypoplastic alae nasi, open mouth, and large ears. De novo KIAA2022 mutations (p.Q705X in Patient 1, p.R322X in Patient 2) were detected by targeted sequencing and confirmed by Sanger sequencing. KIAA2022 mutations and alterations have been reported in only four families with nonsyndromic ID and epilepsy. KIAA2022 is highly expressed in the fetal and adult brain and plays a crucial role in neuronal development. These additional patients support the evidence that KIAA2022 is a causative gene for XLID.


Asunto(s)
Trastorno Autístico/genética , Hipotiroidismo Congénito/genética , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Mutación Missense , Proteínas del Tejido Nervioso/genética , Alelos , Trastorno Autístico/diagnóstico , Trastorno Autístico/patología , Preescolar , Hipotiroidismo Congénito/diagnóstico , Hipotiroidismo Congénito/patología , Expresión Génica , Genes Ligados a X , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/patología , Linaje , Fenotipo
16.
Pediatr Int ; 57(2): 324-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25868953

RESUMEN

Myoclonus-dystonia syndrome (MDS) is a rare autosomal-dominant movement disorder characterized by brief, frequently alcohol-responsive myoclonic jerks that begin in childhood or early adolescence, caused by mutations in the ε-sarcoglycan gene (SGCE). The patient was a 6-year-old boy. At 2 years 8 months, he had abnormal movement when he ran due to dystonia of his left leg. At 3 years 5 months, he exhibited dystonia and myoclonic movement of his arms when eating. Myoclonus was likely to develop when he felt anxiety or exhaustion. Genomic DNA showed a heterozygous mutation in SGCE (c.109 + 1 G > T). His father and uncle with the same mutation also experienced milder dystonia or myoclonic movements. SGCE mutation can cause a broad range of clinical symptoms between and within families. We should consider MDS as a differential diagnosis for patients with paroxysmal walking abnormalities and/or myoclonic movements.


Asunto(s)
Trastornos Distónicos/diagnóstico , Mutación , Sarcoglicanos/genética , Pueblo Asiatico , Niño , Trastornos Distónicos/genética , Humanos , Masculino , Linaje
17.
No To Hattatsu ; 47(1): 49-52, 2015 Jan.
Artículo en Japonés | MEDLINE | ID: mdl-25803912

RESUMEN

Creatine transporter deficiency (CRTR-D) is an X-linked disorder characterized by hypotonia, developmental delay, and seizures. We report the third Japanese family with CRTR-D. The proband was an 8-year-old boy who presented with hypotonia, severe intellectual disability and two episodes of seizures associated with/without fever. Among 7 siblings (4 males, 3 females), the eldest brother had severe intellectual disability, epilepsy, and sudden death at 17 years of age, while 18-year-old third elder brother had severe intellectual disability, autism, and drug-resistant epilepsy. The proband's urinary creatine/creatinine ratio was increased. A reduced creatine peak on brain magnetic resonance spectroscopy and a known pathogenic mutation in the SLC6A8 gene (c.1661 C > T;p.Pro554Leu) confirmed the diagnosis of CRTR-D. The same mutation was found in the third elder brother. Their mother was a heterozygote. Symptoms of CRTR-D are non-specific. Urinary creatine/creatinine ratio should be measured in patients with hypotonia, developmental delay, seizure and autism whose family history indicates an X-linked inheritance.


Asunto(s)
Encefalopatías Metabólicas Innatas/genética , Creatina/deficiencia , Creatina/orina , Creatinina/orina , Epilepsia/etiología , Discapacidad Intelectual Ligada al Cromosoma X/genética , Mutación/genética , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/deficiencia , Adolescente , Encefalopatías Metabólicas Innatas/complicaciones , Encefalopatías Metabólicas Innatas/diagnóstico , Encefalopatías Metabólicas Innatas/patología , Niño , Creatina/genética , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/complicaciones , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Discapacidad Intelectual Ligada al Cromosoma X/patología , Linaje , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética
18.
J Hum Genet ; 59(7): 408-10, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24898829

RESUMEN

In clinical practice, it is important to diagnose the carrier state of female patients with X-linked diseases for genetic counseling to calculate the recurrent risk of offspring. Because some X-linked diseases show high rates of gonadal mosaicism, this diagnosis is sometimes difficult, when there are few offspring in a family and no mutation is detected in the maternal genomic DNA. Here, we report two male siblings with ATR-X syndrome carrying an intragenic deletion of 78.6 kb involving exons 2-5 out of the 35 exons in the ATRX, as revealed by PCR amplification of these exons. The mother was expected to be an obligate carrier, but we could not confirm her as a mutation carrier by quantitative PCR (qPCR) for the exons. However, we identified the breakpoint of ATRX, and qPCR with breakpoint-specific primers revealed gonosomal mosaicism, with a relative frequency of the mutation of <1% in genomic DNA of her peripheral blood. For these obligate carriers of X-linked disease, we should aggressively investigate the maternal genomic status, not only because her genetic condition is important for estimating the recurrent risk of her offspring but also because a diagnosis of her gonosomal mosaicism can render negligible the possibility that her female siblings are carriers. We should reconfirm that a female who has a risk of being a carrier has a gonosomal or somatic mutation, even if she is an obligate carrier or apparently harbors a mutation.


Asunto(s)
ADN Helicasas/genética , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Discapacidad Intelectual Ligada al Cromosoma X/genética , Mosaicismo , Proteínas Nucleares/genética , Eliminación de Secuencia , Talasemia alfa/diagnóstico , Talasemia alfa/genética , Alelos , Niño , Preescolar , Puntos de Rotura del Cromosoma , Análisis Mutacional de ADN , Exones , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Linaje , Fenotipo , Hermanos , Translocación Genética , Proteína Nuclear Ligada al Cromosoma X
19.
Ann Neurol ; 73(1): 48-57, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23225343

RESUMEN

OBJECTIVE: Recently, COL4A1 mutations have been reported in porencephaly and other cerebral vascular diseases, often associated with ocular, renal, and muscular features. In this study, we aimed to clarify the phenotypic spectrum and incidence of COL4A1 mutations. METHODS: We screened for COL4A1 mutations in 61 patients with porencephaly and 10 patients with schizencephaly, which may be similarly caused by disturbed vascular supply leading to cerebral degeneration, but can be distinguished depending on time of insult. RESULTS: COL4A1 mutations were identified in 15 patients (21%, 10 mutations in porencephaly and 5 mutations in schizencephaly), who showed a variety of associated findings, including intracranial calcification, focal cortical dysplasia, pontocerebellar atrophy, ocular abnormalities, myopathy, elevated serum creatine kinase levels, and hemolytic anemia. Mutations include 10 missense, a nonsense, a frameshift, and 3 splice site mutations. Five mutations were confirmed as de novo events. One mutation was cosegregated with familial porencephaly, and 2 mutations were inherited from asymptomatic parents. Aberrant splicing was demonstrated by reverse transcriptase polymerase chain reaction analyses in 2 patients with splice site mutations. INTERPRETATION: Our study first confirmed that COL4A1 mutations are associated with schizencephaly and hemolytic anemia. Based on the finding that COL4A1 mutations were frequent in patients with porencephaly and schizencephaly, genetic testing for COL4A1 should be considered for children with these conditions.


Asunto(s)
Encefalopatías/genética , Colágeno Tipo IV/genética , Hemiplejía/genética , Malformaciones del Desarrollo Cortical/genética , Mutación/genética , Fenotipo , Anemia Hemolítica/genética , Anemia Hemolítica/patología , Encefalopatías/patología , Niño , Preescolar , Colágeno Tipo IV/deficiencia , Hemiplejía/patología , Humanos , Lactante , Malformaciones del Desarrollo Cortical/patología , Porencefalia
20.
Am J Med Genet A ; 164A(11): 2873-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25099823

RESUMEN

Angelman syndrome (AS) is characterized by severe intellectual disability with ataxia, epilepsy, and behavioral uniqueness. The underlining molecular deficit is the absence of the maternal copy of the imprinted UBE3A gene due to maternal deletions, which is observed in 70-75% of cases, and can be detected using fluorescent in situ hybridization (FISH) of the UBE3A region. Only a few familial AS cases have been reported with a complete deletion of UBE3A. Here, we report on siblings with AS caused by a microdeletion of 15q11.2-q12 encompassing UBE3A at the breakpoint of an inversion at 15q11.2 and 15q26.1. Karyotyping revealed an inversion of 15q, and FISH revealed the deletion of the UBE3A region. Array comparative genomic hybridization (CGH) demonstrated a 467 kb deletion at 15q11.2-q12, encompassing only UBE3A, SNORD115, and PAR1, and a 53 kb deletion at 15q26.1, encompassing a part of SLCO3A1. Their mother had a normal karyotype and array CGH detected no deletion of 15q11.2-q12, so we assumed gonadal mosaicism. This report describes a rare type of familial AS detected using the D15S10 FISH test.


Asunto(s)
Síndrome de Angelman/genética , Puntos de Rotura del Cromosoma , Inversión Cromosómica , Cromosomas Humanos Par 15 , Eliminación de Gen , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/diagnóstico , Niño , Preescolar , Mapeo Cromosómico , Hibridación Genómica Comparativa , Facies , Humanos , Hibridación Fluorescente in Situ , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Hermanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA